Single Cell Regulatory Landscape of the Mouse Kidney Highlights Cellular Differentiation Programs and Disease Targets

Total Page:16

File Type:pdf, Size:1020Kb

Single Cell Regulatory Landscape of the Mouse Kidney Highlights Cellular Differentiation Programs and Disease Targets ARTICLE https://doi.org/10.1038/s41467-021-22266-1 OPEN Single cell regulatory landscape of the mouse kidney highlights cellular differentiation programs and disease targets Zhen Miao 1,2,3,8, Michael S. Balzer 1,2,8, Ziyuan Ma 1,2,8, Hongbo Liu1,2, Junnan Wu 1,2, Rojesh Shrestha 1,2, Tamas Aranyi1,2, Amy Kwan4, Ayano Kondo 4, Marco Pontoglio 5, Junhyong Kim6, ✉ Mingyao Li 7, Klaus H. Kaestner2,4 & Katalin Susztak 1,2,4 1234567890():,; Determining the epigenetic program that generates unique cell types in the kidney is critical for understanding cell-type heterogeneity during tissue homeostasis and injury response. Here, we profile open chromatin and gene expression in developing and adult mouse kidneys at single cell resolution. We show critical reliance of gene expression on distal regulatory elements (enhancers). We reveal key cell type-specific transcription factors and major gene- regulatory circuits for kidney cells. Dynamic chromatin and expression changes during nephron progenitor differentiation demonstrates that podocyte commitment occurs early and is associated with sustained Foxl1 expression. Renal tubule cells follow a more complex differentiation, where Hfn4a is associated with proximal and Tfap2b with distal fate. Mapping single nucleotide variants associated with human kidney disease implicates critical cell types, developmental stages, genes, and regulatory mechanisms. The single cell multi-omics atlas reveals key chromatin remodeling events and gene expression dynamics associated with kidney development. 1 Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA. 2 Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA. 3 Graduate Group in Genomics and Computational Biology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA. 4 Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA. 5 Epigenetics and Development Laboratory, Université de Paris Inserm U1151/CNRS UMR 8253, Institut Necker Enfants Malades, Paris, France. 6 Department of Biology, University of Pennsylvania, Philadelphia, PA, USA. 7 Department of Epidemiology and Biostatistics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA. 8These authors contributed equally: Zhen ✉ Miao, Michael S. Balzer, Ziyuan Ma. email: [email protected] NATURE COMMUNICATIONS | (2021) 12:2277 | https://doi.org/10.1038/s41467-021-22266-1 | www.nature.com/naturecommunications 1 ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-021-22266-1 he mammalian kidney maintains fluid, electrolyte, and seq data, susztaklab.com/developing_adult_kidney/scRNA/ for Tmetabolite balance of the body and plays an essential role scRNA-seq data, and susztaklab.com/developing_adult_kidney/igv/ in blood pressure regulation and red blood cell home- for IGV view of peak tracks). Using this atlas, we produce an ostasis. The human kidney makes roughly 180 liters of primary epigenome-based classification of developing and mature cells and filtrate each day that is then reabsorbed and modified by a long defined cell type-specific regulatory networks. We also investigate tubule segment. To perform this highly choreographed and key TFs and cell–cell interactions associated with developmental sophisticated function, the kidney contains close to 20 highly cellular transitions. Finally, we use the single cell open chromatin specialized epithelial cells. The renal glomerulus acts as a 60 kD information to pinpoint putative target genes and cell types of size-selective filter. The proximal part of the tubules is responsible several chronic kidney disease noncoding genome-wide association for reclaiming more than 70% of the primary filtrate, which is study (GWAS) loci. done via unregulated active and passive paracellular transport1, while the loop of Henle plays an important role in concentrating the urine. The distal convoluted tubule is critical for regulated Results sodium reabsorption. The last segment of kidney tubules is the Single cell accessible chromatin landscape of the developing collecting duct, where the final concentration of the urine is and adult mouse kidneys. To characterize the accessible chro- determined via regulation of water channels, acid, or base matin landscape of the developing and adult mouse kidneys at secretion. Understanding the development of these diverse cell single cell resolution, we performed single nuclei ATAC-seq types in the kidney is essential to understand kidney homeostasis, (snATAC-seq) on kidneys of mice on postnatal day 0 (P0), at 3 disease, and regeneration. and 8 weeks (P21 and P56) of age (Fig. 1a). In mice at birth, the The mammalian kidney develops from the intermediate meso- nephron progenitors are still present and nephrons are formed derm via a complex interaction between the ureteric bud and the until around day 2113,18. In parallel, we also performed bulk metanephric mesenchyme2. In the mouse kidney, Six2 marks the (whole kidney) ATAC-seq analysis at matched developmental self-renewing nephron progenitor population3. The nephron pro- stages. Following sequencing, we aggregated all high-quality genitors commit and undergo a mesenchymal-to-epithelial trans- mapped reads in each sample irrespective of barcode. The com- formation giving rise to the renal vesicle3. The renal vesicle then bined snATAC-seq dataset from all samples showed the expected undergoes segmentation and elongation, giving rise to epithelia insert size periodicity (Supplementary Fig. 1a) with a strong from the podocytes to the distal convoluted tubules, while the enrichment of signal at Transcription Start Sites (TSS), indicating ureteric bud becomes the collecting duct. Unbiased and high data quality (Supplementary Fig. 1b). The snATAC-seq data hypothesis-driven studies have highlighted critical stages and dri- showed high concordance with the bulk ATAC data (Spearman vers of early kidney development4, that have been essential for the correlation coefficient >0.84 between matched stages, see development of an in vitro kidney organoid differentiation “Methods” section, Supplementary Fig. 1c). protocols5–7 . However, cells in organoids are still poorly differ- We next revealed cell type annotations from the open entiated, improving cellular differentiation and maturation of these chromatin information. After conducting stringent filtering of structures remains a major challenge8. Thus, the understanding of the number of unique fragments, promoter ratio and mitochon- late kidney development, especially the cell type-specificdriver dria ratio (see “Methods” section, Supplementary Fig. 2a), we kept transcription factors (TFs) is of great importance9–11.Alterationin 28,316 cells across the samples (Fig. 1b). Cells were then clustered Wnt, Notch, Bmp, and Egf signaling significantly impacts cellular using SnapATAC19, which binned the whole genome into 5 kb differentiation, but only a handful of TFs that directly drive the regions to address the sparsity of the data (see “Methods” differentiation of distinct segments have been identified, such as section). Prior to clustering, we used Harmony20, an iterative Pou3f3, Lhx1, Irx2, Foxc2,andMafb12. Further understanding of batch correction method, to correct for variability across samples the terminal differentiation program could aid the understanding (Supplementary Fig. 2b). Using batch-corrected low dimensional of kidney disease development. embeddings, we retained 13 clusters, all of which had consistent While single cell RNA sequencing (scRNA-seq) has improved representation across the number of peaks, samples and read our understanding of kidney development in mice and depth profiles (Figs. 1b and S2c, d). humans9,10,13,14, it provides limited information of TFs, which To determine the cell types represented by each cluster, we are usually expressed at low levels. Equally difficult is to under- examined chromatin accessibility around the TSS and gene body stand how genes are regulated from scRNA-seq data alone. regions of the known cell type-specific marker genes21.Basedonthe Chromatin state profiles, on the other hand, determine the gene accessibility of the known marker genes, we identified clusters expression potential and can pinpoint the availability of TF representing nephron progenitors, endothelial cells, podocytes, binding sites. Together with gene expression, open chromatin proximal tubule segment 1 and segment 3 cells, loop of Henle, distal profiles can define the gene regulatory logic, which is the fun- convoluted tubule, connecting tubule, collecting duct principal cells, damental element of cell identity. However, there is a scarcity of collecting duct intercalated cells, stromal, and immune cells. Fig. 1d open chromatin information by Assay for Transposase-Accessible and Supplmentary Fig. 3) show chromatin accessibility information Chromatin using sequencing (ATAC-seq) or chromatin immu- for key cell type marker genes, such as Uncx and Cited1 for nephron noprecipitation (ChIP) data by ChIP-seq related to kidney post- progenitors, Nphs1 and Nphs2 for podocytes, Akr1c21 for both natal development. In addition, epigenetic changes observed in segments of proximal tubules, Slc34a1 and Slc5a2 for proximal bulk analyses mostly represent changes in cell composition, rather convoluted tubules (PCT), Kap for proximal straight tubules (PST), than cell type-specific
Recommended publications
  • Osr1 Maintains Renal Progenitors and Regulates Podocyte Development by Promoting Wnt2ba Through Antagonism of Hand2
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.21.423845; this version posted December 22, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. osr1 maintains renal progenitors and regulates podocyte development by promoting wnt2ba through antagonism of hand2 Bridgette E. Drummond1, Brooke E. Chambers1, Hannah M. Wesselman1, Marisa N. Ulrich1, Gary F. Gerlach1, Paul T. Kroeger1, Ignaty Leshchiner2, Wolfram Goessling2, and Rebecca A. Wingert1* 1Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, University of Notre Dame, Notre Dame, 46556, USA 2Brigham and Women's Hospital, Genetics and Gastroenterology Division, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA *Corresponding author Keywords: zebrafish, kidney, nephron, podocyte, segmentation, osr1, wnt2ba, hand2 Abbreviations: chronic kidney disease (CKD), N-ethyl-N-nitrosourea (ENU), days post fertilization (dpf), hours post fertilization (hpf), whole mount in situ hybridization (WISH), flourescent in situ hybridization (FISH), immunofluorescence (IF), immunocytochemistry (ICC), intermediate mesoderm (IM), congenital anomalies of the kidney and urinary tract (CAKUT), amino acid (aa), Zebrafish International Research Center (ZIRC), capped RNA (cRNA), somite stage (ss), wild-type (WT), LIM homeobox 1a (lhx1a), paired box 2a (pax2a), wilms tumor 1a (wt1a), wilms tumor 1b (wt1b), nephrosis 1, congenital Finnish type (nephrin) (nphs1), nephrosis 2, idiopathic, steroid-resistant (podocin)(nphs2), odd-skipped related transcription factor 1 (osr1), cadherin 17 (cdh17), odd-skipped related transcription factor 2 (osr2), wingless-type MMTV integration site family, member 2Ba (wnt2ba) and wingless-type MMTV integration site family, member 2Bb (wnt2bb), heart and neural crest derivatives expressed 2 (hand2), myosin light chain (myl7).
    [Show full text]
  • Role of Estrogen Receptor Beta and the Isoflavone Genistein
    WCP2018 OR28-3 Oral session White-to-brown adipose differentiation: role of estrogen receptor beta and the isoflavone genistein Alessandra Bitto, Federica Mannino, Natasha Irrera, Giovanni Pallio, Domenica Altavilla, Francesco Squadrito Clinical and experimental medicine, University of Messina, Italy The two types of fat cells in mammals brown and white have different functions. White adipose tissue (WAT) stores excess energy in the form of triglyceride and releases free fatty acids during caloric deficiency. Brown adipose tissue (BAT) on the other hand can dissipate energy through thermogenesis. Genistein can have an effect on energy expenditure UCP (uncoupling protein) expression and protect against the obesogenic effect of a high calorie diet. The effect of genistein in inducing white-to-brown transdifferentiation was investigated in 3T3-L1 cells differentiated into white adipocytes with a specific medium (DMEM 10% calf serum 1% penicillin/streptomycin 500 uM 3isobutyl1 methylxanthine 10ug/ml insulin 250 nM dexmethasone 8 ug/ml biotin and 4 ug/ml pantothenic acid). Fully differentiated white adipocytes were treated after 10 days with different genistein doses (10-50-100-200 uM) for 24-48h or left untreated. Two specific ER-beta and PPAR-gamma receptor inhibitors were also used to understand if genistein effects are mediated by the estrogen or the PPAR receptor. Also a CRISPR/Cas9 approach was used to delete either ER-beta or PPAR-gamma to clarify which receptor is involved in genistein action. Intracellular lipid accumulation was determined by oil-red-O staining after 24 and 48hours of treatment. The expression of UCP1 estrogen receptor alpha and beta PPARalpha and gamma DIO2 (Type II iodothyronine deiodinase) PRDM16 (PR domain containing 16) and CIDEA (cell death inducing DNA fragmentation factor) were evaluated by qPCR after 24 and 48hours of genistein treatment.
    [Show full text]
  • TRANSCRIPTIONAL REGULATION of Hur in RENAL STRESS
    TRANSCRIPTIONAL REGULATION OF HuR IN RENAL STRESS DISSERTATION Presented in Partial Fulfillment of the Requirements for the Degree Doctor of Philosophy in the Graduate School of The Ohio State University By Sudha Suman Govindaraju Graduate Program in Biochemistry The Ohio State University 2014 Dissertation Committee: Dr. Beth S. Lee, Ph.D., Advisor Dr. Kathleen Boris-Lawrie, Ph.D. Dr. Sissy M. Jhiang, Ph.D. Dr. Arthur R. Strauch, Ph.D Abstract HuR is a ubiquitously expressed RNA-binding protein that affects the post- transcriptional life of thousands of cellular mRNAs by regulating transcript stability and translation. HuR can post-transcriptionally regulate gene expression and modulate cellular responses to stress, differentiation, proliferation, apoptosis, senescence, inflammation, and the immune response. It is an important mediator of survival during cellular stress, but when inappropriately expressed, can promote oncogenic transformation. Not surprisingly, the expression of HuR itself is tightly regulated at multiple transcriptional and post-transcriptional levels. Previous studies demonstrated the existence of two alternate HuR transcripts that differ in their 5’ untranslated regions and have markedly different translatabilities. These forms were also found to be reciprocally expressed following cellular stress in kidney proximal tubule cell lines, and the shorter, more readily translatable variant was shown to be regulated by Smad 1/5/8 pathway and bone morphogenetic protein-7 (BMP-7) signaling. In this study, the factors that promote transcription of the longer alternate form were identified. NF-κB was shown to be important for expression of the long HuR mRNA, as was a newly identified region with potential for binding the Sp/KLF families of transcription factors.
    [Show full text]
  • Id Proteins Promote a Cancer Stem Cell Phenotype in Triple Negative Breast
    bioRxiv preprint doi: https://doi.org/10.1101/497313; this version posted March 11, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license. 1 Id proteins promote a cancer stem cell phenotype in triple negative 2 breast cancer via negative regulation of Robo1 3 Wee S. Teo1,2, Holly Holliday1,2#, Nitheesh Karthikeyan3#, Aurélie S. Cazet1,2, Daniel L. 4 Roden1,2, Kate Harvey1, Christina Valbirk Konrad1, Reshma Murali3, Binitha Anu Varghese3, 5 Archana P. T.3,4, Chia-Ling Chan1,2, Andrea McFarland1, Simon Junankar1,2, Sunny Ye1, 6 Jessica Yang1, Iva Nikolic1,2, Jaynish S. Shah5, Laura A. Baker1,2, Ewan K.A. Millar1,6,7,8, 7 Mathew J. Naylor1,2,10, Christopher J. Ormandy1,2, Sunil R. Lakhani11, Warren Kaplan1,12, 8 Albert S. Mellick13,14, Sandra A. O’Toole1,9, Alexander Swarbrick1,2*, Radhika Nair1,2,3* 9 Affiliations 10 1Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia 11 2St Vincent’s Clinical School, Faculty of Medicine, UNSW Sydney, New South Wales, 12 Australia 13 3 Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Kerala, India 14 4Manipal Academy of Higher Education, Manipal, Karnataka, India 15 5Centenary Institute, The University of Sydney, New South Wales, Australia 16 6 Department of Anatomical Pathology, NSW Health Pathology, St George Hospital, 17 Kogarah, NSW, Australia 18 7 School of Medical Sciences, UNSW Sydney, Kensington NSW, Australia.
    [Show full text]
  • Human Stem Cells from Single Blastomeres Reveal Pathways of Embryonic Or Trophoblast Fate Specification Tamara Zdravkovic1,2,3,4,5,‡, Kristopher L
    © 2015. Published by The Company of Biologists Ltd | Development (2015) 142, 4010-4025 doi:10.1242/dev.122846 STEM CELLS AND REGENERATION RESEARCH ARTICLE Human stem cells from single blastomeres reveal pathways of embryonic or trophoblast fate specification Tamara Zdravkovic1,2,3,4,5,‡, Kristopher L. Nazor6,‡, Nicholas Larocque1,2,3,4,5, Matthew Gormley1,2,3,4,5, Matthew Donne1,2,3,7, Nathan Hunkapillar1,2,3,4,5, Gnanaratnam Giritharan8, Harold S. Bernstein4,9, Grace Wei4,10, Matthias Hebrok10, Xianmin Zeng11, Olga Genbacev1,2,3,4,5, Aras Mattis4,12, Michael T. McMaster4,5,13, Ana Krtolica8,*, Diana Valbuena14, Carlos Simón14, Louise C. Laurent6,15, Jeanne F. Loring6 and Susan J. Fisher1,2,3,4,5,7,§ ABSTRACT INTRODUCTION For many reasons, relatively little is known about human Mechanisms of initial cell fate decisions differ among species. To gain preimplantation development. The small number of cells makes insights into lineage allocation in humans, we derived ten human embryos of any species difficult to study. In humans, the technical embryonic stem cell lines (designated UCSFB1-10) from single difficulties are compounded by other challenges. Genetic variation blastomeres of four 8-cell embryos and one 12-cell embryo from a among individuals could contribute to developmental differences, a single couple. Compared with numerous conventional lines from well-appreciated phenomenon in the mouse (Dackor et al., 2009), blastocysts, they had unique gene expression and DNA methylation which is difficult to assess in humans owing to the limited patterns that were, in part, indicative of trophoblast competence. At a availability of embryos that are donated for research.
    [Show full text]
  • Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model
    Downloaded from http://www.jimmunol.org/ by guest on September 25, 2021 T + is online at: average * The Journal of Immunology , 34 of which you can access for free at: 2016; 197:1477-1488; Prepublished online 1 July from submission to initial decision 4 weeks from acceptance to publication 2016; doi: 10.4049/jimmunol.1600589 http://www.jimmunol.org/content/197/4/1477 Molecular Profile of Tumor-Specific CD8 Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A. Waugh, Sonia M. Leach, Brandon L. Moore, Tullia C. Bruno, Jonathan D. Buhrman and Jill E. Slansky J Immunol cites 95 articles Submit online. Every submission reviewed by practicing scientists ? is published twice each month by Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts http://jimmunol.org/subscription Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html http://www.jimmunol.org/content/suppl/2016/07/01/jimmunol.160058 9.DCSupplemental This article http://www.jimmunol.org/content/197/4/1477.full#ref-list-1 Information about subscribing to The JI No Triage! Fast Publication! Rapid Reviews! 30 days* Why • • • Material References Permissions Email Alerts Subscription Supplementary The Journal of Immunology The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2016 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. This information is current as of September 25, 2021. The Journal of Immunology Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A.
    [Show full text]
  • Podocyte Specific Knockdown of Klf15 in Podocin-Cre Klf15flox/Flox Mice Was Confirmed
    SUPPLEMENTARY FIGURE LEGENDS Supplementary Figure 1: Podocyte specific knockdown of Klf15 in Podocin-Cre Klf15flox/flox mice was confirmed. (A) Primary glomerular epithelial cells (PGECs) were isolated from 12-week old Podocin-Cre Klf15flox/flox and Podocin-Cre Klf15+/+ mice and cultured at 37°C for 1 week. Real-time PCR was performed for Nephrin, Podocin, Synaptopodin, and Wt1 mRNA expression (n=6, ***p<0.001, Mann-Whitney test). (B) Real- time PCR was performed for Klf15 mRNA expression (n=6, *p<0.05, Mann-Whitney test). (C) Protein was also extracted and western blot analysis for Klf15 was performed. The representative blot of three independent experiments is shown in the top panel. The bottom panel shows the quantification of Klf15 by densitometry (n=3, *p<0.05, Mann-Whitney test). (D) Immunofluorescence staining for Klf15 and Wt1 was performed in 12-week old Podocin-Cre Klf15flox/flox and Podocin-Cre Klf15+/+ mice. Representative images from four mice in each group are shown in the left panel (X 20). Arrows show colocalization of Klf15 and Wt1. Arrowheads show a lack of colocalization. Asterisk demonstrates nonspecific Wt1 staining. “R” represents autofluorescence from RBCs. In the right panel, a total of 30 glomeruli were selected in each mouse and quantification of Klf15 staining in the podocytes was determined by the ratio of Klf15+ and Wt1+ cells to Wt1+ cells (n=6 mice, **p<0.01, unpaired t test). Supplementary Figure 2: LPS treated Podocin-Cre Klf15flox/flox mice exhibit a lack of recovery in proteinaceous casts and tubular dilatation after DEX administration.
    [Show full text]
  • Letters to the Editor
    LETTERS TO THE EDITOR The closely related rare and severe acute myeloid leukemias carrying EVI1 or PRDM16 rearrangements share singular biological features In a recent issue of Haematologica , Matsuo et al .1 pinpoint the pejorative effect of EVI1 overexpression in 18 acute myeloid leukemias (AML) with MLL rearrangements. However, EVI1 overexpression has also been reported in patients with translocations involving chromosome 3 and the EVI1 gene. 2,3 Because of the poor prognosis associated to these anomalies, it is important to investigate them at an early stage in order to adapt patient management. Indeed, previous reports 4-6 and the 2008 WHO classification 7 indi - cate that EVI1-rearranged (EVI1-r) AML display typical fea - tures, such as absence of thrombopenia, atypical megakary - Figure 1. Algorithm for the suspicion of EVI1 and PRDM16 AMLs. ocytes and multilineage dysplasia 2-4 which can be detected by current diagnostic reference methods. In this line, we compared a cohort of 17 EVI1-r AML, aged between 8 and 79-years old (median 54 years) to 1822 other cases of AML months. diagnosed in the same laboratory over 14 years. At diagno - This study consolidates the unusual base-line character - sis, there were similar hemoglobin levels or white blood istics and clinical features of EVI1-r AML cases. Moreover, cell counts in both groups. Median platelet counts were it indicates a very low rate of MPO expression in EVI1-r 9 9 123x10 /L, higher than 100x10 /L in 53% of EVI1-r AML AML patients. It is interesting to note that relationships patients, compared to 25% in the control AML population have been reported between EVI1 expression and MPO (P=0.02).
    [Show full text]
  • Pluripotency Factors Regulate Definitive Endoderm Specification Through Eomesodermin
    Downloaded from genesdev.cshlp.org on September 23, 2021 - Published by Cold Spring Harbor Laboratory Press Pluripotency factors regulate definitive endoderm specification through eomesodermin Adrian Kee Keong Teo,1,2 Sebastian J. Arnold,3 Matthew W.B. Trotter,1 Stephanie Brown,1 Lay Teng Ang,1 Zhenzhi Chng,1,2 Elizabeth J. Robertson,4 N. Ray Dunn,2,5 and Ludovic Vallier1,5,6 1Laboratory for Regenerative Medicine, University of Cambridge, Cambridge CB2 0SZ, United Kingdom; 2Institute of Medical Biology, A*STAR (Agency for Science, Technology, and Research), Singapore 138648; 3Renal Department, Centre for Clinical Research, University Medical Centre, 79106 Freiburg, Germany; 4Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom Understanding the molecular mechanisms controlling early cell fate decisions in mammals is a major objective toward the development of robust methods for the differentiation of human pluripotent stem cells into clinically relevant cell types. Here, we used human embryonic stem cells and mouse epiblast stem cells to study specification of definitive endoderm in vitro. Using a combination of whole-genome expression and chromatin immunoprecipitation (ChIP) deep sequencing (ChIP-seq) analyses, we established an hierarchy of transcription factors regulating endoderm specification. Importantly, the pluripotency factors NANOG, OCT4, and SOX2 have an essential function in this network by actively directing differentiation. Indeed, these transcription factors control the expression of EOMESODERMIN (EOMES), which marks the onset of endoderm specification. In turn, EOMES interacts with SMAD2/3 to initiate the transcriptional network governing endoderm formation. Together, these results provide for the first time a comprehensive molecular model connecting the transition from pluripotency to endoderm specification during mammalian development.
    [Show full text]
  • Cited1 (NM 001276466) Mouse Tagged ORF Clone Product Data
    OriGene Technologies, Inc. 9620 Medical Center Drive, Ste 200 Rockville, MD 20850, US Phone: +1-888-267-4436 [email protected] EU: [email protected] CN: [email protected] Product datasheet for MR228487 Cited1 (NM_001276466) Mouse Tagged ORF Clone Product data: Product Type: Expression Plasmids Product Name: Cited1 (NM_001276466) Mouse Tagged ORF Clone Tag: Myc-DDK Symbol: Cited1 Synonyms: AI316840; AU019144; Msg1 Vector: pCMV6-Entry (PS100001) E. coli Selection: Kanamycin (25 ug/mL) Cell Selection: Neomycin ORF Nucleotide >MR228487 representing NM_001276466 Sequence: Red=Cloning site Blue=ORF Green=Tags(s) TTTTGTAATACGACTCACTATAGGGCGGCCGGGAATTCGTCGACTGGATCCGGTACCGAGGAGATCTGCC GCCGCGATCGCC ATGCCAACTATGTCGAGGCCTGCACTTGATGTCAAGGGTGGCACCACCTCTGGGAAGGAGGATGCCAACC AGGAGATGAACTCTCTGGCCTACTCCAACCTTGGAGTGAAGGATCGCAAGGCAGTGACTGTCCTGCACTA CCCCGGGGTCACCGCAAATGGAGCCAAAGCCAACGGAGTTCCCACTAGCTCCTCTGGATCGACATCTCCA ATAGGCTCTCCTACTGCCACCCCTTCTTCCAAACCCCCATCCTTCAACCTGCATCCTACCCCTCACCTGA TGGCCAGCATGCAGCTTCAGAAGCTTAATAGCCAGTACCAAGGGGCTGCGGCTACTGCTGCTGCTGCCCT CACTGGTGCAGGCCTACCAGGGGAGGAAGAGCCCATGCAAAACTGGGTCACCGCCCCTCTGGTAGTGGGA GGGTCTCCGGGATCTGTCTCTCCTCCTGCTGGTGCCCAGAGCCCTGCTCTCATTGATTCTGACCCGGTGG ATGAGGAGGTGCTGATGTCTCTGGTGGTTGAATTGGGGCTAGACCGAGCCAATGAGCTTCCCGAGCTGTG GCTGGGGCAGAATGAGTTTGATTTCACTGCAGATTTTCCCTCTGGCTGC ACGCGTACGCGGCCGCTCGAGCAGAAACTCATCTCAGAAGAGGATCTGGCAGCAAATGATATCCTGGATT ACAAGGATGACGACGATAAGGTTTAA Protein Sequence: >MR228487 representing NM_001276466 Red=Cloning site Green=Tags(s) MPTMSRPALDVKGGTTSGKEDANQEMNSLAYSNLGVKDRKAVTVLHYPGVTANGAKANGVPTSSSGSTSP
    [Show full text]
  • Variable Clinical Expression in Patients with a Germline MEN1 Disease Gene Mutation: Clues to a Genotype–Phenotype Correlation
    CLINICS 2012;67(S1):49-56 DOI:10.6061/clinics/2012(Sup01)10 REVIEW Variable clinical expression in patients with a germline MEN1 disease gene mutation: clues to a genotype–phenotype correlation Cornelis J. Lips,I Koen M. Dreijerink,I Jo W. Ho¨ ppenerII I University Medical Center Utrecht, Department of Internal Medicine & Endocrinology, Utrecht, The Netherlands. II University Medical Center Utrecht, Department of Metabolic & Endocrine Diseases, Utrecht, The Netherlands. Multiple endocrine neoplasia type 1 is an inherited endocrine tumor syndrome, predominantly characterized by tumors of the parathyroid glands, gastroenteropancreatic tumors, pituitary adenomas, adrenal adenomas, and neuroendocrine tumors of the thymus, lungs or stomach. Multiple endocrine neoplasia type 1 is caused by germline mutations of the multiple endocrine neoplasia type 1 tumor suppressor gene. The initial germline mutation, loss of the wild-type allele, and modifying genetic and possibly epigenetic and environmental events eventually result in multiple endocrine neoplasia type 1 tumors. Our understanding of the function of the multiple endocrine neoplasia type 1 gene product, menin, has increased significantly over the years. However, to date, no clear genotype– phenotype correlation has been established. In this review we discuss reports on exceptional clinical presentations of multiple endocrine neoplasia type 1, which may provide more insight into the pathogenesis of this disorder and offer clues for a possible genotype–phenotype correlation. KEYWORDS: Multiple Endocrine Neoplasia type 1; MEN1; Menin; Genotype–Phenotype Correlation; Clinical Expression. Lips CJ, Dreijerink KM, Ho¨ ppener JW. Variable clinical expression in patients with a germline MEN1 disease gene mutation: clues to a genotype– phenotype correlation. Clinics. 2012;67(S1):49-56.
    [Show full text]
  • Supplemental Materials ZNF281 Enhances Cardiac Reprogramming
    Supplemental Materials ZNF281 enhances cardiac reprogramming by modulating cardiac and inflammatory gene expression Huanyu Zhou, Maria Gabriela Morales, Hisayuki Hashimoto, Matthew E. Dickson, Kunhua Song, Wenduo Ye, Min S. Kim, Hanspeter Niederstrasser, Zhaoning Wang, Beibei Chen, Bruce A. Posner, Rhonda Bassel-Duby and Eric N. Olson Supplemental Table 1; related to Figure 1. Supplemental Table 2; related to Figure 1. Supplemental Table 3; related to the “quantitative mRNA measurement” in Materials and Methods section. Supplemental Table 4; related to the “ChIP-seq, gene ontology and pathway analysis” and “RNA-seq” and gene ontology analysis” in Materials and Methods section. Supplemental Figure S1; related to Figure 1. Supplemental Figure S2; related to Figure 2. Supplemental Figure S3; related to Figure 3. Supplemental Figure S4; related to Figure 4. Supplemental Figure S5; related to Figure 6. Supplemental Table S1. Genes included in human retroviral ORF cDNA library. Gene Gene Gene Gene Gene Gene Gene Gene Symbol Symbol Symbol Symbol Symbol Symbol Symbol Symbol AATF BMP8A CEBPE CTNNB1 ESR2 GDF3 HOXA5 IL17D ADIPOQ BRPF1 CEBPG CUX1 ESRRA GDF6 HOXA6 IL17F ADNP BRPF3 CERS1 CX3CL1 ETS1 GIN1 HOXA7 IL18 AEBP1 BUD31 CERS2 CXCL10 ETS2 GLIS3 HOXB1 IL19 AFF4 C17ORF77 CERS4 CXCL11 ETV3 GMEB1 HOXB13 IL1A AHR C1QTNF4 CFL2 CXCL12 ETV7 GPBP1 HOXB5 IL1B AIMP1 C21ORF66 CHIA CXCL13 FAM3B GPER HOXB6 IL1F3 ALS2CR8 CBFA2T2 CIR1 CXCL14 FAM3D GPI HOXB7 IL1F5 ALX1 CBFA2T3 CITED1 CXCL16 FASLG GREM1 HOXB9 IL1F6 ARGFX CBFB CITED2 CXCL3 FBLN1 GREM2 HOXC4 IL1F7
    [Show full text]